Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 130(6): 1284-93, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-21469141

ABSTRACT

Susceptibility contrast magnetic resonance imaging (MRI), utilising ultrasmall superparamagnetic iron oxide (USPIO) particles, was evaluated for the quantitation of vessel size index (Rv, µm), a weighted average measure of tumour blood vessel calibre, and fractional tumour blood volume (fBV, %), in orthotopically propagated murine PC3 prostate tumour xenografts. Tumour vascular architecture was assessed in vivo by MRI prior to and 24 hr after treatment with 200 mg/kg of the vascular disrupting agent ZD6126. A Bayesian hierarchical model (BHM) was used to reduce the uncertainty associated with quantitation of Rv and fBV. Quantitative histological analyses of the uptake of Hoechst 33342 for perfused vasculature, and haematoxylin and eosin staining for necrosis, were also performed to qualify the MRI data. A relatively large median Rv of 40.3 µm (90% confidence interval (CI90) = 37.4, 44.0 µm) and a high fBV of 5.4% (CI90 = 5.3, 5.5%) were determined in control tumours, which agreed with histologically determined vessel size index. Treatment with ZD6126 significantly (p < 0.01) reduced tumour Rv (34.2 µm, CI90 = 31.2, 38.0 µm) and fBV (3.9%, CI90 = 3.8, 4.1%), which were validated against histologically determined significant reductions in perfusion and vessel size, and increased necrosis. Together these data (i) highlight the use of a BHM to optimise the inferential power available from susceptibility contrast MRI data, (ii) provide strong evaluation and qualification of R(v) and fBV as non-invasive imaging biomarkers of tumour vascular morphology, (iii) reveal the presence of a different vascular phenotype and (iv) demonstrate that ZD6126 exhibits good anti-vascular activity against orthotopic prostate tumours.


Subject(s)
Magnetic Resonance Imaging/methods , Prostatic Neoplasms/blood supply , Animals , Bayes Theorem , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Necrosis/drug therapy , Necrosis/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Organophosphorus Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Transplantation, Heterologous
2.
J Mol Histol ; 42(4): 341-54, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21732047

ABSTRACT

Hypoxia is thought to be critical in regulating physiological processes within the female reproductive system, including ovulation, composition of the fluid in the oviductal/uterine lumens and ovarian follicle development. This study examined the localisation of exogenous (pimonidazole) and endogenous [hypoxia inducible factor 1α and 2α (HIF1α, -2α), glucose transporter type 1 (GLUT1) and carbonic anhydrase 9 (CAIX)] hypoxia-related antigens within the oviduct and uterus of the rat reproductive tract. The extent to which each endogenous antigen co-compartmentalised with pimonidazole was also assessed. Female Wistar Furth rats (n = 10) were injected intraperitoneally with pimonidazole (60 mg/kg) 1 h prior to death. Reproductive tissues were removed immediately following death and fixed in 4% paraformaldehyde before being embedded in paraffin. Serial sections were cut (6-7 µm thick) and antigens of interest identified using standard immunohistochemical procedures. The mucosal epithelia of the ampulla, isthmus and uterus were immunopositive for pimonidazole in most sections. Co-compartmentalisation of pimonidazole with HIF1α was only expressed in the mucosa of the uterus whilst co-compartmentalisation with HIF2α was observed in the mucosa of the ampulla, isthmus and uterus. Both GLUT1 and CAIX were co-compartmentalised with pimonidazole in mucosa of the isthmus and uterus. This study confirms that mucosal regions of the rat oviduct and uterus frequently experience severe hypoxia and there are compartment specific variations in expression of endogenous hypoxia-related antigens, including the HIF isoforms. The latter observation may relate to target gene specificity of HIF isoforms or perhaps HIF2α's responsiveness to non-hypoxic stimuli such as hypoglycaemia independently of HIF1α.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Endometrium/metabolism , Fallopian Tubes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endometrium/cytology , Epithelium/metabolism , Fallopian Tubes/cytology , Female , Glucose Transporter Type 1/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mucous Membrane/cytology , Mucous Membrane/metabolism , Myometrium/cytology , Myometrium/metabolism , Rats
3.
Magn Reson Med ; 64(3): 914-21, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20806382

ABSTRACT

Although the biasing of R(2)* estimates by assuming magnitude MR data to be normally distributed has been described, the effect on changes in R(2)* (DeltaR(2)*), such as induced by a paramagnetic contrast agent, has not been reported. In this study, two versions of a novel Bayesian maximum a posteriori approach for estimating DeltaR(2)* are described and evaluated: one that assumes normally distributed data and the other, Rice-distributed data. The approach enables the robust, voxelwise determination of the uncertainty in DeltaR(2)* estimates and provides a useful statistical framework for quantifying the probability that a pixel has been significantly enhanced. This technique was evaluated in vivo, using ultrasmall superparamagnetic iron oxide particles in orthotopic murine prostate tumors. It is shown that assuming magnitude data to be normally distributed causes DeltaR(2)* to be underestimated when signal-to-noise ratio is modest. However, the biasing effect is less than is found in R(2)* estimates, implying that the simplifying assumption of normally distributed noise is more justifiable when evaluating DeltaR(2)* compared with when evaluating precontrast R(2)* values.


Subject(s)
Algorithms , Artificial Intelligence , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Pattern Recognition, Automated/methods , Prostatic Neoplasms/pathology , Animals , Bayes Theorem , Cell Line, Tumor , Humans , Image Enhancement/methods , Male , Mice , Mice, Nude , Reproducibility of Results , Sensitivity and Specificity
4.
Radiology ; 254(1): 110-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20032145

ABSTRACT

PURPOSE: To investigate relationships between magnetic resonance (MR) imaging measurements of R2* and carbogen-induced DeltaR2* in vivo with subsequent histologic assessment of grade, hypoxia, fibrosis, and necrosis in a chemically induced rat mammary tumor model. MATERIALS AND METHODS: All experiments were performed in accordance with the local ethics review panel, the UK Home Office Animals Scientific Procedures Act of 1986, and the UK Co-ordinating Committee on Cancer Research guidelines. Of 30 rats injected with N-methyl-N-nitrosourea, 17 developed mammary tumors. Prior to MR imaging, rats were administered pimonidazole. Tumor R2* was then quantified while the host first breathed air and then carbogen (95% O(2), 5% CO(2); n = 16). Tumor sections were subsequently stained for pimonidazole, sirius red, cytokeratin 14, and hematoxylin-eosin for quantitative assessment of hypoxia, fibrosis, malignancy, and necrosis, respectively, and graded according to the Scarff-Bloom-Richardson scale. Linear regression analysis was used to identify any correlates of the MR imaging data with histologic data. RESULTS: Tumors exhibited wide heterogeneity in the magnitude of carbogen-induced reduction in R2*, an emerging imaging biomarker of fractional blood volume. Significant correlations were found between pimonidazole adduct formation and both baseline tumor R2* (r = -0.54, P = .03) and carbogen-induced DeltaR2* (r = 0.56, P = .02), demonstrating that tumors with a larger fractional blood volume were less hypoxic. There was also a significant correlation between pimonidazole and sirius red staining (r = 0.76, P < .01), indicating that more fibrotic tumors were also more hypoxic. There were no correlations of R2* with grade. CONCLUSION: In this model of breast cancer, baseline tumor R2* and carbogen-induced DeltaR2* are predictive imaging biomarkers for hypoxia and primarily determined by blood volume.


Subject(s)
Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/chemically induced , Animals , Biomarkers, Tumor/metabolism , Carbon Dioxide/pharmacology , Female , Fibrosis/pathology , Hypoxia/pathology , Keratin-14/metabolism , Linear Models , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Necrosis/pathology , Nitroimidazoles/pharmacology , Oxygen/pharmacology , Radiation-Sensitizing Agents/pharmacology , Rats , Rats, Wistar
5.
Magn Reson Med ; 62(2): 420-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19353661

ABSTRACT

The least-squares algorithm is known to bias apparent diffusion coefficient (ADC) values estimated from magnitude MR data, although this effect is commonly assumed to be negligible. In this study the effect of this bias on tumor ADC estimates was evaluated in vivo and was shown to introduce a consistent and significant underestimation of ADC, relative to those given by a robust maximum likelihood approach (on average, a 23.4 +/- 12% underestimation). Monte Carlo simulations revealed that the magnitude of the bias increased with ADC and decreasing signal-to-noise ratio (SNR). In vivo, this resulted in a much-reduced ability to resolve necrotic regions from surrounding viable tumor tissue compared with a maximum likelihood approach. This has significant implications for the evaluation of diffusion MR data in vivo, in particular in heterogeneous tumor tissue, when evaluating bi- and multiexponential tumor diffusion models for the modeling of data acquired with larger b-values (b > 1000 s/mm(2)) and for data with modest SNR. Use of a robust approach to modeling magnitude MR data from tumors is therefore recommended over the least-squares approach when evaluating data from heterogeneous tumors.


Subject(s)
Algorithms , Diffusion Magnetic Resonance Imaging/methods , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Neoplasms/diagnosis , Reproducibility of Results , Sensitivity and Specificity
6.
Int J Radiat Oncol Biol Phys ; 71(5): 1470-6, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18538948

ABSTRACT

PURPOSE: Vascular disrupting agents are anticancer agents that typically produce a cytostatic tumor response. Vessel size index magnetic resonance imaging (MRI) allows for the estimation of the fractional blood volume (fBV) and blood vessel size (Rv). We assessed whether the vessel size index parameters provided imaging biomarkers for detecting early tumor response to a vascular disrupting agent. METHODS AND MATERIALS: GH3 prolactinomas were grown subcutaneously in 12 rats. Vessel size index MRI was performed with Sinerem, an ultrasmall superparamagnetic iron oxide intravascular contrast agent, to determine the tumor fBV and Rv. MRI was performed before and at 24 h after treatment with either the vascular disrupting agent, 5,6-dimethylxanthenone 4-acetic acid (DMXAA) (n = 6) or with the drug vehicle (n = 6). After treatment, the tumors were analyzed histologically and correlates with the MRI findings sought. RESULTS: Histogram analysis showed non-normal distributions of Rv and fBV. The 25th percentiles of the fBV and Rv were significantly reduced (p < 0.01) after treatment with DMXAA, with an increase in the regions of low-measured fBV. For the treated and control tumors, the fraction of tumor with an fBV of < or =1% correlated with the histologically determined percentage of necrosis (r = 0.77, p < 0.005). The fraction of tumor with an fBV of < or =1% in treated tumors was significantly increased compared with before treatment (p < 0.05) and with that in the controls (p < 0.05). CONCLUSION: The vessel size index results were consistent with the known action of DMXAA to cause vascular collapse, with histogram analysis of the fBV providing the most sensitive indicator of response. In particular, the parameter, the fraction of tumor with an fBV of < or =1% is a potential biomarker that correlates with the histopathologic measure of tumor necrosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Blood Volume Determination/methods , Prolactinoma/blood supply , Prolactinoma/drug therapy , Xanthones/therapeutic use , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Blood Volume , Contrast Media , Dextrans , Female , Ferrosoferric Oxide , Iron , Magnetic Resonance Imaging , Magnetite Nanoparticles , Organ Size , Oxides , Prolactinoma/pathology , Prolactinoma/physiopathology , Rats , Rats, Inbred WF , Regional Blood Flow
7.
Int J Radiat Oncol Biol Phys ; 69(4): 1238-45, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17967313

ABSTRACT

PURPOSE: To investigate the use of the transverse magnetic resonance imaging (MRI) relaxation rate R(2)(*) (s(-1)) as a biomarker of tumor vascular response to monitor vascular disrupting agent (VDA) therapy. METHODS AND MATERIALS: Multigradient echo MRI was used to quantify R(2)(*) in rat GH3 prolactinomas. R(2)(*) is a sensitive index of deoxyhemoglobin in the blood and can therefore be used to give an index of tissue oxygenation. Tumor R(2)(*) was measured before and up to 35 min after treatment, and 24 h after treatment with either 350 mg/kg 5,6-dimethylxanthenone-4-acetic acid (DMXAA) or 100 mg/kg combretastatin-A4-phosphate (CA4P). After acquisition of the MRI data, functional tumor blood vessels remaining after VDA treatment were quantified using fluorescence microscopy of the perfusion marker Hoechst 33342. RESULTS: DMXAA induced a transient, significant (p < 0.05) increase in tumor R(2)(*) 7 min after treatment, whereas CA4P induced no significant changes in tumor R(2)(*) over the first 35 min. Twenty-four hours after treatment, some DMXAA-treated tumors demonstrated a decrease in R(2)(*), but overall, reduction in R(2)(*) was not significant for this cohort. Tumors treated with CA4P showed a significant (p < 0.05) reduction in R(2)(*) 24 h after treatment. The degree of Hoechst 33342 uptake was associated with the degree of R(2)(*) reduction at 24 h for both agents. CONCLUSIONS: The reduction in tumor R(2)(*) or deoxyhemoglobin levels 24 h after VDA treatment was a result of reduced blood volume caused by prolonged vascular collapse. Our results suggest that DMXAA was less effective than CA4P in this rat tumor model.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Magnetic Resonance Imaging/methods , Neoplasms/blood supply , Neoplasms/drug therapy , Stilbenes/therapeutic use , Xanthones/therapeutic use , Animals , Biomarkers, Tumor/metabolism , Female , Hemoglobins/metabolism , Neoplasms/blood , Neovascularization, Pathologic/drug therapy , Rats , Rats, Inbred WF
8.
Neoplasia ; 8(3): 199-206, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16611413

ABSTRACT

The dose-dependent effects of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) on rat GH3 prolactinomas were investigated in vivo. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used to assess tumor blood flow/permeability pretreatment and 24 hours posttreatment with 0, 100, 200, or 350 mg/kg DMXAA. DCE-MRI data were analyzed using K(trans) and the integrated area under the gadolinium time curve (IAUGC) as response biomarkers. High-performance liquid chromatography (HPLC) was used to determine the plasma concentration of the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) following treatment to provide an index of increased vessel permeability and vascular damage. Finally, tumor necrosis was assessed by grading hematoxylin and eosin-stained sections cut from the same tumors investigated by MRI. Both tumor K(trans) and IAUGC were significantly reduced 24 hours posttreatment with 350 mg/kg DMXAA only, with no evidence of dose response. HPLC demonstrated a significant increase in plasma 5-HIAA 24 hours posttreatment with 200 and 350 mg/kg DMXAA. Histologic analysis revealed some evidence of tumor necrosis following treatment with 100 or 200 mg/kg DMXAA, reaching significance with 350 mg/kg DMXAA. The absence of any reduction in K(trans) or IAUGC following treatment with 200 mg/kg, despite a significant increase in 5-HIAA, raises concerns about the utility of established DCE-MRI biomarkers to assess tumor response to DMXAA.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Indoles/blood , Magnetic Resonance Imaging , Neovascularization, Pathologic/drug therapy , Pituitary Neoplasms/pathology , Prolactinoma/blood supply , Xanthones/therapeutic use , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Area Under Curve , Biomarkers , Capillary Permeability , Cell Line, Tumor/transplantation , Contrast Media , Dose-Response Relationship, Drug , Female , Gadolinium DTPA , Necrosis , Neoplasm Transplantation , Prolactinoma/drug therapy , Prolactinoma/pathology , Rats , Rats, Inbred WF , Subcutaneous Tissue , Xanthones/pharmacokinetics , Xanthones/pharmacology
9.
Clin Cancer Res ; 11(10): 3705-13, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897567

ABSTRACT

PURPOSE: To use (31)P and (1)H magnetic resonance spectroscopy (MRS) to assess changes in tumor metabolic profile in vivo in response to 5,6-dimethylxanthenone-4-acetic acid (DMXAA) with a view to identifying biomarkers associated with tumor dose response. EXPERIMENTAL DESIGN: In vivo (31)P and (1)H MRS measurements of (a) tumor bioenergetics [beta-nucleoside triphosphate/inorganic phosphate (beta-NTP/Pi)], (b) the membrane-associated phosphodiesters and phosphomonoesters (PDE/PME), (c) choline (mmol/L), and (d) lactate/water ratio were made on murine HT29 colon carcinoma xenografts pretreatment and 6 or 24 hours posttreatment with increasing doses of DMXAA. Following in vivo MRS, the tumors were excised and used for high-resolution (31)P and (1)H MRS of extracts to provide validation of the in vivo MRS data, histologic analysis of necrosis, and high-performance liquid chromatography. RESULTS: Both beta-NTP/Pi and PDE/PME decreased in a dose-dependent manner 6 hours posttreatment with DMXAA, with significant decreases in beta-NTP/Pi with 15 mg/kg (P < 0.001) and 21 mg/kg (P < 0.01). A significant decrease in total choline in vivo was found 24 hours posttreatment with 21 mg/kg DMXAA (P < 0.05); this was associated with a significant reduction in the concentration of the membrane degradation products glycerophosphoethanolamine and glycerophosphocholine measured in tissue extracts (P < 0.05). CONCLUSIONS: The reduction in tumor energetics and membrane turnover is consistent with the vascular-disrupting activity of DMXAA. (31)P MRS revealed tumor response to DMXAA at doses below the maximum tolerated dose for mice. Both (31)P and (1)H MRS provide biomarkers of tumor response to DMXAA that could be used in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Xanthones/pharmacology , Animals , Carcinoma/metabolism , Cell Membrane/metabolism , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , Energy Metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Nude , Neovascularization, Pathologic , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...